Assessment of Systemic Delivery of rAAVrh74.MHCK7.micro-dystrophin in Children With Duchenne Muscular Dystrophy: A Nonrandomized Controlled Trial

Jerry R Mendell, Zarife Sahenk, Kelly Lehman, Carrie Nease, Linda P Lowes, Natalie F Miller, Megan A Iammarino, Lindsay N Alfano, Amanda Nicholl, Samiah Al-Zaidy, Sarah Lewis, Kathleen Church, Richard Shell, Linda H Cripe, Rachael A Potter, Danielle A Griffin, Eric Pozsgai, Ashish Dugar, Mark Hogan, Louise R Rodino-Klapac, Jerry R Mendell, Zarife Sahenk, Kelly Lehman, Carrie Nease, Linda P Lowes, Natalie F Miller, Megan A Iammarino, Lindsay N Alfano, Amanda Nicholl, Samiah Al-Zaidy, Sarah Lewis, Kathleen Church, Richard Shell, Linda H Cripe, Rachael A Potter, Danielle A Griffin, Eric Pozsgai, Ashish Dugar, Mark Hogan, Louise R Rodino-Klapac

Abstract

Importance: Micro-dystrophin gene transfer shows promise for treating patients with Duchenne muscular dystrophy (DMD) using recombinant adeno-associated virus serotype rh74 (rAAVrh74) and codon-optimized human micro-dystrophin driven by a skeletal and cardiac muscle-specific promoter with enhanced cardiac expression (MHCK7).

Objective: To identify the 1-year safety and tolerability of intravenous rAAVrh74.MHCK7.micro-dystrophin in patients with DMD.

Design, setting, and participants: This open-label, phase 1/2a nonrandomized controlled trial was conducted at the Nationwide Children's Hospital in Columbus, Ohio. It began on November 2, 2017, with a planned duration of follow-up of 3 years, ending in March 2021. The first 4 patients who met eligibility criteria were enrolled, consisting of ambulatory male children with DMD without preexisting AAVrh74 antibodies and a stable corticosteroid dose (≥12 weeks).

Interventions: A single dose of 2.0 × 1014 vg/kg rAAVrh74.MHCK7.micro-dystrophin was infused through a peripheral limb vein. Daily prednisolone, 1 mg/kg, started 1 day before gene delivery (30-day taper after infusion).

Main outcomes and measures: Safety was the primary outcome. Secondary outcomes included micro-dystrophin expression by Western blot and immunohistochemistry. Functional outcomes measured by North Star Ambulatory Assessment (NSAA) and serum creatine kinase were exploratory outcomes.

Results: Four patients were included (mean [SD] age at enrollment, 4.8 [1.0] years). All adverse events (n = 53) were considered mild (33 [62%]) or moderate (20 [38%]), and no serious adverse events occurred. Eighteen adverse events were considered treatment related, the most common of which was vomiting (9 of 18 events [50%]). Three patients had transiently elevated γ-glutamyltransferase, which resolved with corticosteroids. At 12 weeks, immunohistochemistry of gastrocnemius muscle biopsy specimens revealed robust transgene expression in all patients, with a mean of 81.2% of muscle fibers expressing micro-dystrophin with a mean intensity of 96% at the sarcolemma. Western blot showed a mean expression of 74.3% without fat or fibrosis adjustment and 95.8% with adjustment. All patients had confirmed vector transduction and showed functional improvement of NSAA scores and reduced creatine kinase levels (posttreatment vs baseline) that were maintained for 1 year.

Conclusions and relevance: This trial showed rAAVrh74.MHCK7.micro-dystrophin to be well tolerated and have minimal adverse events; the safe delivery of micro-dystrophin transgene; the robust expression and correct localization of micro-dystrophin protein; and improvements in creatine kinase levels and NSAA scores. These findings suggest that rAAVrh74.MHCK7.micro-dystrophin can provide functional improvement that is greater than that observed under standard of care.

Trial registration: ClinicalTrials.gov Identifier: NCT03375164.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Mendell reported receiving grants from Parent Project Muscular Dystrophy; receiving personal fees from Sarepta Therapeutics Inc., and Nationwide Children's Hospital outside the submitted work; and holding a pending patent to micro-dystrophin cassette for gene therapy and an issued patent to rAAV.SGCA delivery isolated limb infusion. Dr Sahenk reported receiving grants from Sarepta Therapeutics Inc during the conduct of the study. Ms Lehman reported receiving grants from Sarepta Therapeutics Inc. during the conduct of the study. Ms Nease reported receiving grants from Nationwide Children's Hospital during the conduct of the study. Ms Nicholl reported receiving grants from Nationwide Children's Hospital during the conduct of the study. Dr Al-Zaidy reported receiving consulting fees from AveXis Inc outside the submitted work. Dr Lewis reported being an employee of Sarepta Therapeutics Inc. during the conduct of the study. Dr Potter reported receiving other from Sarepta Therapeutics Inc. during the conduct of the . Ms Griffin reported receiving other from Sarepta Therapeutics Inc. during the conduct of the study. Dr Pozsgai reported receiving other from Sarepta Therapeutics Inc. during the conduct of the study and outside the submitted work. Dr Dugar reported being an employee of Sarepta Therapeutics Inc. Dr Rodino-Klapac reported receiving personal fees from Myonexus Therapeutics outside the submitted work, holding a patent (pending, licensed, and with royalties paid) to adeno-associated virus delivery of muscle-specific micro-dystrophin to treat patients with muscular dystrophy, and being an employee of Sarepta Therapeutics Inc. No other disclosures were reported.

Figures

Figure 1.. Study Flow Diagram
Figure 1.. Study Flow Diagram
Figure 2.. Lack of Vector-Associated Toxic Effects…
Figure 2.. Lack of Vector-Associated Toxic Effects by Histopathological Assessments in 4 Patients
A, Hematoxylin-eosin staining (original magnification ×20) shows unremarkable alterations in central nucleation (yellow arrowheads) of muscle fibers before and after treatment. Baseline (pretreatment) biopsy results show levels of necrosis (white arrowheads), stages of degeneration or regeneration, and increased connective tissue (black arrowheads). B, Picrosirius red staining shows a reduction in fibrosis after treatment. C, Fibrosis in tissue samples was measured by the percentage of collagen accumulation in tissue section. The error bars refer to the SD.
Figure 3.. Systemic Administration of rAAVrh74.MHCK7.micro-dystrophin
Figure 3.. Systemic Administration of rAAVrh74.MHCK7.micro-dystrophin
A, Frozen biopsied tissue sections of the gastrocnemius muscle were processed and stained with DYS3 antibody to detect micro-dystrophin. B, Micro-dystrophin compared with full-length protein was seen in 4 patients with Duchenne muscular dystrophy (DMD) before and after treatment (day 90) with rAAVrh74.MHCK7.micro-dystrophin. α-Actinin was used as a loading control, and sample from a patient was included as a negative control. NC indicates normal control. aPatient 4 had a positive immunoreactive band that was above the upper limit of quantification and was diluted 1 to 4 before loading in the representative gel.

References

    1. Mendell JR, Lloyd-Puryear M. Report of MDA muscle disease symposium on newborn screening for Duchenne muscular dystrophy. Muscle Nerve. 2013;48(1):21-26. doi:10.1002/mus.23810
    1. Mendell JR, Shilling C, Leslie ND, et al. . Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann Neurol. 2012;71(3):304-313. doi:10.1002/ana.23528
    1. Bello L, Morgenroth LP, Gordish-Dressman H, Hoffman EP, McDonald CM, Cirak S; CINRG investigators . DMD genotypes and loss of ambulation in the CINRG Duchenne Natural History Study. Neurology. 2016;87(4):401-409. doi:10.1212/WNL.0000000000002891
    1. Birnkrant DJ, Bushby K, Bann CM, et al. ; DMD Care Considerations Working Group . Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. 2018;17(4):347-361. doi:10.1016/S1474-4422(18)30025-5
    1. Birnkrant DJ, Bushby K, Bann CM, et al. ; DMD Care Considerations Working Group . Diagnosis and management of Duchenne muscular dystrophy, part 3: primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 2018;17(5):445-455. doi:10.1016/S1474-4422(18)30026-7
    1. Henricson EK, Abresch RT, Cnaan A, et al. ; CINRG Investigators . The cooperative international neuromuscular research group Duchenne natural history study: glucocorticoid treatment preserves clinically meaningful functional milestones and reduces rate of disease progression as measured by manual muscle testing and other commonly used clinical trial outcome measures. Muscle Nerve. 2013;48(1):55-67. doi:10.1002/mus.23808
    1. Liu D, Ahmet A, Ward L, et al. . A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol. 2013;9(1):30. doi:10.1186/1710-1492-9-30
    1. Rice JB, White AG, Scarpati LM, Wan G, Nelson WW. Long-term systemic corticosteroid exposure: a systematic literature review. Clin Ther. 2017;39(11):2216-2229. doi:10.1016/j.clinthera.2017.09.011
    1. Khan N, Eliopoulos H, Han L, et al. ; Eteplirsen Investigators and the CINRG DNHS Investigators . Eteplirsen treatment attenuates respiratory decline in ambulatory and non-ambulatory patients with Duchenne muscular dystrophy. J Neuromuscul Dis. 2019;6(2):213-225. doi:10.3233/JND-180351
    1. Aartsma-Rus A, Verschuuren JJGM, Campion GV, van Ommen GB, van Deutekom JCT. Overview on DMD exon skipping. Methods Mol Biol. 2012;867:97-116. doi:10.1007/978-1-61779-767-5_7
    1. Charleston JS, Schnell FJ, Dworzak J, et al. . Eteplirsen treatment for Duchenne muscular dystrophy: exon skipping and dystrophin production. Neurology. 2018;90(24):e2146-e2154. doi:10.1212/WNL.0000000000005680
    1. Cirak S, Arechavala-Gomeza V, Guglieri M, et al. . Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet. 2011;378(9791):595-605. doi:10.1016/S0140-6736(11)60756-3
    1. Mendell JR, Al-Zaidy S, Shell R, et al. . Single-dose gene-replacement therapy for spinal muscular atrophy. N Engl J Med. 2017;377(18):1713-1722. doi:10.1056/NEJMoa1706198
    1. Lowes LP, Alfano LN, Arnold WD, et al. . Impact of age and motor function in a phase 1/2A study of infants with SMA type 1 receiving single-dose gene replacement therapy. Pediatr Neurol. 2019;98:39-45. doi:10.1016/j.pediatrneurol.2019.05.005
    1. Al-Zaidy SA, Kolb SJ, Lowes L, et al. . AVXS-101 (onasemnogene abeparvovec) for SMA1: comparative study with a prospective natural history cohort. J Neuromuscul Dis. 2019;6(3):307-317. doi:10.3233/JND-190403
    1. Duan D. Systemic AAV micro-dystrophin gene therapy for Duchenne muscular dystrophy. Mol Ther. 2018;26(10):2337-2356. doi:10.1016/j.ymthe.2018.07.011
    1. Wells DJ, Wells KE, Asante EA, et al. . Expression of human full-length and minidystrophin in transgenic mdx mice: implications for gene therapy of Duchenne muscular dystrophy. Hum Mol Genet. 1995;4(8):1245-1250. doi:10.1093/hmg/4.8.1245
    1. England SB, Nicholson LV, Johnson MA, et al. . Very mild muscular dystrophy associated with the deletion of 46% of dystrophin. Nature. 1990;343(6254):180-182. doi:10.1038/343180a0
    1. Zhao J, Kodippili K, Yue Y, et al. . Dystrophin contains multiple independent membrane-binding domains. Hum Mol Genet. 2016;25(17):3647-3653. doi:10.1093/hmg/ddw210
    1. Nelson DM, Lindsay A, Judge LM, et al. . Variable rescue of microtubule and physiological phenotypes in mdx muscle expressing different miniaturized dystrophins. Hum Mol Genet. 2018;27(12):2090-2100. doi:10.1093/hmg/ddy113
    1. Harper SQ, Hauser MA, DelloRusso C, et al. . Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy. Nat Med. 2002;8(3):253-261. doi:10.1038/nm0302-253
    1. Suzuki A, Yoshida M, Yamamoto H, Ozawa E. Glycoprotein-binding site of dystrophin is confined to the cysteine-rich domain and the first half of the carboxy-terminal domain. FEBS Lett. 1992;308(2):154-160. doi:10.1016/0014-5793(92)81265-N
    1. Salva MZ, Himeda CL, Tai PW, et al. . Design of tissue-specific regulatory cassettes for high-level rAAV-mediated expression in skeletal and cardiac muscle. Mol Ther. 2007;15(2):320-329. doi:10.1038/sj.mt.6300027
    1. Rodino-Klapac LR, Janssen PM, Montgomery CL, et al. . A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy. J Transl Med. 2007;5:45. doi:10.1186/1479-5876-5-45
    1. Mendell JR, Rodino-Klapac LR, Rosales-Quintero X, et al. . Limb-girdle muscular dystrophy type 2D gene therapy restores alpha-sarcoglycan and associated proteins. Ann Neurol. 2009;66(3):290-297. doi:10.1002/ana.21732
    1. Potter RA, Griffin DA, Heller KN, et al. . Dose escalation study of systemically delivered AAVrh74.MHCK7.micro-dystrophin in the mdx mouse model of DMD. Data presented at: 2018 American Society of Gene & Cell Therapy Annual Meeting; May 16-18, 2018; Chicago, Illinois.
    1. Dixon JR. The International Conference on Harmonization Good Clinical Practice guideline. Qual Assur. 1998;6(2):65-74. doi:10.1080/105294199277860
    1. Mendell JR, Campbell K, Rodino-Klapac L, et al. . Dystrophin immunity in Duchenne’s muscular dystrophy. N Engl J Med. 2010;363(15):1429-1437. doi:10.1056/NEJMoa1000228
    1. Alfano LN, Miller NF, Berry KM, et al. . The 100-meter timed test: normative data in healthy males and comparative pilot outcome data for use in Duchenne muscular dystrophy clinical trials. Neuromuscul Disord. 2017;27(5):452-457. doi:10.1016/j.nmd.2017.02.007
    1. Escolar DM, Hache LP, Clemens PR, et al. . Randomized, blinded trial of weekend vs daily prednisone in Duchenne muscular dystrophy. Neurology. 2011;77(5):444-452. doi:10.1212/WNL.0b013e318227b164
    1. Sondergaard PC, Griffin DA, Pozsgai ER, et al. . AAV.dysferlin overlap vectors restore function in dysferlinopathy animal models. Ann Clin Transl Neurol. 2015;2(3):256-270. doi:10.1002/acn3.172
    1. Schnepp BC, Jensen RL, Chen CL, Johnson PR, Clark KR. Characterization of adeno-associated virus genomes isolated from human tissues. J Virol. 2005;79(23):14793-14803. doi:10.1128/JVI.79.23.14793-14803.2005
    1. Tarnopolsky MA, Pearce E, Smith K, Lach B. Suction-modified Bergström muscle biopsy technique: experience with 13,500 procedures. Muscle Nerve. 2011;43(5):717-725. doi:10.1002/mus.21945
    1. Stevens W. Good clinical laboratory practice (GCLP): the need for a hybrid of good laboratory practice and good clinical practice guidelines/standards for medical testing laboratories conducting clinical trials in developing countries. Qual Assur. 2003;10(2):83-89. doi:10.1080/10529410390262727
    1. Schnell FJ, Frank D, Fletcher S, Johnsen RD, Wilton SD. Challenges of interpreting dystrophin content by western blot. US Neurol. 2019;15(1):40-46. doi:10.17925/USN.2019.15.1.40
    1. Silva MC, Meira ZM, Gurgel Giannetti J, et al. . Myocardial delayed enhancement by magnetic resonance imaging in patients with muscular dystrophy. J Am Coll Cardiol. 2007;49(18):1874-1879. doi:10.1016/j.jacc.2006.10.078
    1. Kawel-Boehm N, Maceira A, Valsangiacomo-Buechel ER, et al. . Normal values for cardiovascular magnetic resonance in adults and children. J Cardiovasc Magn Reson. 2015;17:29. doi:10.1186/s12968-015-0111-7
    1. A randomized, double-blind, placebo-controlled study of SRP-9001 for Duchenne muscular dystrophy (DMD). Identifier: NCT03769116. Updated March 30, 2020. Accessed February 12, 2020.
    1. Manno CS, Pierce GF, Arruda VR, et al. . Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12(3):342-347. Published correction appears in Nat Med. 2006;12(5):592.
    1. Nathwani AC, Tuddenham EG, Rangarajan S, et al. . Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med. 2011;365(25):2357-2365. doi:10.1056/NEJMoa1108046
    1. Doriguzzi C, Palmucci L, Mongini T, Chiadò-Piat L, Restagno G, Ferrone M. Exercise intolerance and recurrent myoglobinuria as the only expression of Xp21 Becker type muscular dystrophy. J Neurol. 1993;240(5):269-271. doi:10.1007/BF00838159
    1. Ricotti V, Ridout DA, Pane M, et al. ; UK NorthStar Clinical Network . The NorthStar Ambulatory Assessment in Duchenne muscular dystrophy: considerations for the design of clinical trials. J Neurol Neurosurg Psychiatry. 2016;87(2):149-155. doi:10.1136/jnnp-2014-309405
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; PTC124-GD-007-DMD Study Group . The 6-minute walk test and other clinical endpoints in Duchenne muscular dystrophy: reliability, concurrent validity, and minimal clinically important differences from a multicenter study. Muscle Nerve. 2013;48(3):357-368. doi:10.1002/mus.23905
    1. Mazzone E, Vasco G, Sormani MP, et al. . Functional changes in Duchenne muscular dystrophy: a 12-month longitudinal cohort study. Neurology. 2011;77(3):250-256. doi:10.1212/WNL.0b013e318225ab2e
    1. Muntoni F, Domingos J, Manzur AY, et al. ; UK NorthStar Network . Categorising trajectories and individual item changes of the North Star Ambulatory Assessment in patients with Duchenne muscular dystrophy. PLoS One. 2019;14(9):e0221097. doi:10.1371/journal.pone.0221097
    1. Ricotti V, Ridout DA, Scott E, et al. ; NorthStar Clinical Network . Long-term benefits and adverse effects of intermittent versus daily glucocorticoids in boys with Duchenne muscular dystrophy. J Neurol Neurosurg Psychiatry. 2013;84(6):698-705. doi:10.1136/jnnp-2012-303902
    1. Wong B, Signorovitch J, Staunton H, et al. . Estimating clinically meaningful change thresholds in the North Star Ambulatory Assessment (NSAA) and four-stair climb (4SC) in Duchenne muscular dystrophy (DMD). Neuromuscul Disord. 2019;29:S106. doi:10.1016/j.nmd.2019.06.251

Source: PubMed

3
Subskrybuj